1
|
Gallagher LT, LaCroix I, Fields AT, Mitra S, Argabright A, D'Alessandro A, Erickson C, Nunez-Garcia B, Herrera-Rodriguez K, Chou YC, Stocker BW, Ramser BJ, Thielen O, Hallas W, Silliman CC, Kornblith LZ, Cohen MJ. Platelet releasates mitigate the endotheliopathy of trauma. J Trauma Acute Care Surg 2024; 97:738-746. [PMID: 38764145 PMCID: PMC11502277 DOI: 10.1097/ta.0000000000004342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
BACKGROUND Platelets are well known for their roles in hemostasis, but they also play a key role in thromboinflammatory pathways by regulating endothelial health, stimulating angiogenesis, and mediating host defense through both contact dependent and independent signaling. When activated, platelets degranulate releasing multiple active substances. We hypothesized that the soluble environment formed by trauma platelet releasates (TPR) attenuates thromboinflammation via mitigation of trauma induced endothelial permeability and metabolomic reprogramming. METHODS Blood was collected from injured and healthy patients to generate platelet releasates and plasma in parallel. Permeability of endothelial cells when exposed to TPR and plasma (TP) was assessed via resistance measurement by electric cell-substrate impedance sensing (ECIS). Endothelial cells treated with TPR and TP were subjected to mass spectrometry-based metabolomics. RESULTS TP increased endothelial permeability, whereas TPR decreased endothelial permeability when compared with untreated cells. When TP and TPR were mixed ex vivo, TPR mitigated TP-induced permeability, with significant increase in AUC compared with TP alone. Metabolomics of TPR and TP demonstrated disrupted redox reactions and anti-inflammatory mechanisms. CONCLUSION Trauma platelet releasates provide endothelial barrier protection against TP-induced endothelial permeability. Our findings highlight a potential beneficial action of activated platelets on the endothelium in injured patients through disrupted redox reactions and increased antioxidants. Our findings support that soluble signaling from platelet degranulation may mitigate the endotheliopathy of trauma. The clinical implications of this are that activated platelets may prove a promising therapeutic target in the complex integration of thrombosis, endotheliopathy, and inflammation in trauma.
Collapse
Affiliation(s)
- Lauren T Gallagher
- From the Department of Surgery (L.T.G., S.M., B.W.S., B.J.R., O.T., W.H., M.J.C.), Department of Biochemistry and Molecular Genetics (I.L.C., C.E.), University of Colorado, School of Medicine, Aurora, Colorado; Department of Surgery (A.T.F., B.N.-G., K.H.-R., Y.C.C., L.Z.K.), University of California, San Francisco, San Francisco, California; Mass Spectrometry Core Facility (A.A.), University of Colorado, School of Medicine; Department of Biochemistry and Molecular Genetics (A.D'A.), University of Colorado Anschutz Medical Campus, School of Medicine; Vitalant Research Institute, Department of Surgery (C.C.S.), Department of Pediatrics (C.C.S.), University of Colorado, School of Medicine, Aurora, Colorado; and Department of Laboratory Medicine (L.Z.K.), University of California, San Francisco, San Francisco, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Sirtori CR, Castiglione S, Pavanello C. METFORMIN: FROM DIABETES TO CANCER TO PROLONGATION OF LIFE. Pharmacol Res 2024; 208:107367. [PMID: 39191336 DOI: 10.1016/j.phrs.2024.107367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024]
Abstract
The metformin molecule dates back to over a century, but its clinical use started in the '50s. Since then, its use in diabetics has grown constantly, with over 150 million users today. The therapeutic profile also expanded, with improved understanding of novel mechanisms. Metformin has a major activity on insulin resistance, by acting on the insulin receptors and mitochondria, most likely by activation of the adenosine monophosphate-activated kinase. These and associated mechanisms lead to significant lipid lowering and body weight loss. An anti-cancer action has come up in recent years, with mechanisms partly dependent on the mitochondrial activity and also on phosphatidylinositol 3-kinase resistance occurring in some malignant tumors. The potential of metformin to raise life-length is the object of large ongoing studies and of several basic and clinical investigations. The present review article will attempt to investigate the basic mechanisms behind these diverse activities and the potential clinical benefits. Metformin may act on transcriptional activity by histone modification, DNA methylation and miRNAs. An activity on age-associated inflammation (inflammaging) may occur via activation of the nuclear factor erythroid 2 related factor and changes in gut microbiota. A senolytic activity, leading to reduction of cells with the senescent associated secretory phenotype, may be crucial in lifespan prolongation as well as in ancillary properties in age-associated diseases, such as Parkinson's disease. Telomere prolongation may be related to the activity on mitochondrial respiratory factor 1 and on peroxisome gamma proliferator coactivator 1-alpha. Very recent observations on the potential to act on the most severe neurological disorders, such as amyotrophic lateral sclerosis and frontotemporal dementia, have raised considerable hope.
Collapse
Affiliation(s)
- Cesare R Sirtori
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - Sofia Castiglione
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Chiara Pavanello
- Center of Dyslipidemias, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy; Centro E. Grossi Paoletti, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
3
|
Lee JH, Ward KR. Blood failure: traumatic hemorrhage and the interconnections between oxygen debt, endotheliopathy, and coagulopathy. Clin Exp Emerg Med 2024; 11:9-21. [PMID: 38018069 PMCID: PMC11009713 DOI: 10.15441/ceem.23.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 09/28/2023] [Indexed: 11/30/2023] Open
Abstract
This review explores the concept of "blood failure" in traumatic injury, which arises from the interplay of oxygen debt, the endotheliopathy of trauma (EoT), and acute traumatic coagulopathy (ATC). Traumatic hemorrhage leads to the accumulation of oxygen debt, which can further exacerbate hemorrhage by triggering a cascade of events when severe. Such events include EoT, characterized by endothelial glycocalyx damage, and ATC, involving platelet dysfunction, fibrinogen depletion, and dysregulated fibrinolysis. To manage blood failure effectively, a multifaceted approach is crucial. Damage control resuscitation strategies such as use of permissive hypotension, early hemorrhage control, and aggressive transfusion of blood products including whole blood aim to minimize oxygen debt and promote its repayment while addressing endothelial damage and coagulation. Transfusions of red blood cells, plasma, and platelets, as well as the use of tranexamic acid, play key roles in hemostasis and countering ATC. Whole blood, whether fresh or cold-stored, is emerging as a promising option to address multiple needs in traumatic hemorrhage. This review underscores the intricate relationships between oxygen debt, EoT, and ATC and highlights the importance of comprehensive, integrated strategies in the management of traumatic hemorrhage to prevent blood failure. A multidisciplinary approach is essential to address these interconnected factors effectively and to improve patient outcomes.
Collapse
Affiliation(s)
- Jae Hyuk Lee
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kevin R. Ward
- Department of Emergency Medicine, Max Harry Weil Institute for Critical Care Research and Innovation, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Rojas-González DM, Babendreyer A, Ludwig A, Mela P. Analysis of flow-induced transcriptional response and cell alignment of different sources of endothelial cells used in vascular tissue engineering. Sci Rep 2023; 13:14384. [PMID: 37658092 PMCID: PMC10474151 DOI: 10.1038/s41598-023-41247-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 08/23/2023] [Indexed: 09/03/2023] Open
Abstract
Endothelialization of tissue-engineered vascular grafts has proven crucial for implant functionality and thus clinical outcome, however, the choice of endothelial cells (ECs) is often driven by availability rather than by the type of vessel to be replaced. In this work we studied the response to flow of different human ECs with the aim of examining whether their response in vitro is dictated by their original in vivo conditions. Arterial, venous, and microvascular ECs were cultured under shear stress (SS) of 0, 0.3, 3, 1, 10, and 30 dyne/cm2 for 24 h. Regulation of flow-induced marker KLF2 was similar across the different ECs. Upregulation of anti-thrombotic markers, TM and TPA, was mainly seen at higher SS. Cell elongation and alignment was observed for the different ECs at 10 and 30 dyne/cm2 while at lower SS cells maintained a random orientation. Downregulation of pro-inflammatory factors SELE, IL8, and VCAM1 and up-regulation of anti-oxidant markers NQO1 and HO1 was present even at SS for which cell alignment was not observed. Our results evidenced similarities in the response to flow among the different ECs, suggesting that the maintenance of the resting state in vitro is not dictated by the SS typical of the tissue of origin and that absence of flow-induced cell orientation does not necessarily correlate with a pro-inflammatory state of the ECs. These results support the use of ECs from easily accessible sources for in vitro vascular tissue engineering independently from the target vessel.
Collapse
Affiliation(s)
- Diana M Rojas-González
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex) at Center of Biohybrid Medical Systems (CBMS), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany.
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, School of Engineering and Design and Munich Institute of Biomedical Engineering, Technical University of Munich, Boltzmannstr 15, 85748, Garching, Germany.
| |
Collapse
|
5
|
Belvedere R, Novizio N, Eletto D, Porta A, Di Maio U, Petrella A. The Protecting Activity of RIPACUT ®: A New Therapeutic Approach Preserving Epithelial Health Based on the Combination of Iceland Lichen Extract, Silver Salt, and Sodium Hyaluronate. Life (Basel) 2023; 13:life13051088. [PMID: 37240732 DOI: 10.3390/life13051088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Epithelial integrity and function must be maintained in a dynamic healthy equilibrium, keeping unaltered the oxidative and inflammatory conditions and the microbiome of the cutaneous layers. Beside the skin, other mucous membranes can be injured, such as the nasal and anal ones, because of the contact with the external environment. Here, we detected the effects of RIPACUT®, a combination of Iceland lichen extract, silver salt and sodium hyaluronate that individually act in diverse biological ways. The findings we obtained on keratinocytes, nasal and intestinal epithelial cells reveal that this combination showed a marked antioxidant activity, further assessed by the DPPH assay. Additionally, by analyzing the release of the IL-1β, TNF-α and IL-6 cytokines, we proved the anti-inflammatory effect of RIPACUT®. In both cases, the main preserving action was due to Iceland lichen. We also observed a notable antimicrobial activity mediated by the silver compound. These data suggest that RIPACUT® could signify the basis for an attractive pharmacological approach to maintaining healthy epithelial conditions. Interestingly, this may be extended to the nasal and anal areas where it protects against oxidative, inflammatory and infectious insults. Thus, these outcomes encourage the creation of sprays or creams for which sodium hyaluronate can guarantee a surface film-forming effect.
Collapse
Affiliation(s)
- Raffaella Belvedere
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Nunzia Novizio
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Daniela Eletto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Umberto Di Maio
- Shedir Pharma Group Spa, Via Bagnulo 95, 80063 Piano di Sorrento, Italy
| | - Antonello Petrella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| |
Collapse
|
6
|
Bai R, Nguyen TT, Zhou Y, Diao Y, Zhang W. Identification of Antioxidative Peptides Derived from Arthrospira maxima in the Biorefinery Process after Extraction of C-Phycocyanin and Lipids. Mar Drugs 2023; 21:146. [PMID: 36976194 PMCID: PMC10056772 DOI: 10.3390/md21030146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/09/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
Arthrospira maxima has been identified as a sustainable source of rich proteins with diverse functionalities and bioactivities. After extracting C-phycocyanin (C-PC) and lipids in a biorefinery process, the spent biomass still contains a large proportion of proteins with potential for biopeptide production. In this study, the residue was digested using Papain, Alcalase, Trypsin, Protamex 1.6, and Alcalase 2.4 L at different time intervals. The resulting hydrolyzed product with the highest antioxidative activity, evaluated through their scavenging capability of hydroxyl radicals, superoxide anion, 2,2-diphenyl-1-picrylhydrazyl (DPPH), and 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid (ABTS), was selected for further fractionation and purification to isolate and identify biopeptides. Alcalase 2.4 L was found to produce the highest antioxidative hydrolysate product after four-hour hydrolysis. Fractionating this bioactive product using ultrafiltration obtained two fractions with different molecular weights (MW) and antioxidative activity. The low-molecular-weight fraction (LMWF) with MW <3 kDa had higher DPPH scavenging activity with the IC50 value of 2.97 ± 0.33 compared to 3.76 ± 0.15 mg/mL of the high-molecular-weight fraction (HMWF) with MW >3 kDa. Two stronger antioxidative fractions (F-A and F-B) with the respective significant lower IC50 values of 0.83 ± 0.22 and 1.52 ± 0.29 mg/mL were isolated from the LMWF using gel filtration with a Sephadex G-25 column. Based on LC-MS/MS analysis of the F-A, 230 peptides derived from 108 A. maxima proteins were determined. Notably, different antioxidative peptides possessing various bioactivities, including antioxidation, were detected with high predicted scores together with in silico analyses on their stability and toxicity. This study established knowledge and technology to further value-add to the spent A. maxima biomass by optimizing hydrolysis and fraction processes to produce antioxidative peptides with Alcalase 2.4 L after two products already produced in a biorefinery. These bioactive peptides have potential applications in food and nutraceutical products.
Collapse
Affiliation(s)
- Renao Bai
- School of Medicine, Huaqiao University, Quanzhou 362021, China
- Centre for Marine Bioproduct Development, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Trung T. Nguyen
- Centre for Marine Bioproduct Development, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
- Aquaculture Laboratory, College of Science and Engineering, Flinders University, Adelaide, SA 5042, Australia
| | - Yali Zhou
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou 362021, China
| | - Wei Zhang
- Centre for Marine Bioproduct Development, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
- Marine Bioproducts Cooperative Research Centre, Adelaide, SA 5042, Australia
| |
Collapse
|
7
|
Rodríguez JL, Berrios P, Clavo ZM, Marin-Bravo M, Inostroza-Ruiz L, Ramos-Gonzalez M, Quispe-Solano M, Fernández-Alfonso MS, Palomino O, Goya L. Chemical Characterization, Antioxidant Capacity and Anti-Oxidative Stress Potential of South American Fabaceae Desmodium tortuosum. Nutrients 2023; 15:nu15030746. [PMID: 36771451 PMCID: PMC9921092 DOI: 10.3390/nu15030746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
It has been proposed that oxidative stress is a pathogenic mechanism to induce cytotoxicity and to cause cardiovascular and neuronal diseases. At present, natural compounds such as plant extracts have been used to reduce the cytotoxic effects produced by agents that induce oxidative stress. Our study aimed to evaluate the antioxidant and cytoprotective capacity of Desmodium tortuosum (D. tortuosum) extract in the co- and pre-treatment in EA.hy926 and SH-SY5Y cell lines subjected to oxidative stress induced by tert-butylhydroperoxide (t-BOOH). Cell viability, reactive oxygen species (ROS), nitric oxide (NO), caspase 3/7 activity, reduced glutathione (GSH), glutathione peroxidase (GPx), glutathione reductase (GR), and molecular expression of oxidative stress biomarkers (SOD2, NRF2 and NFκB1) and cell death (APAF1, BAX, Caspase3) were all evaluated. It was observed that the D. tortuosum extract, in a dose-dependent manner, was able to reduce the oxidative and cytotoxicity effects induced by t-BOOH, even normalized to a dose of 200 µg/mL, which would be due to the high content of phenolic compounds mainly phenolic acids, flavonoids, carotenoids and other antioxidant compounds. Finally, these results are indicators that the extract of D. tortuosum could be a natural alternative against the cytotoxic exposure to stressful and cytotoxic chemical agents.
Collapse
Affiliation(s)
- José-Luis Rodríguez
- Faculty of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, Lima 15021, Peru
- Faculty of Veterinary, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence:
| | - Paola Berrios
- Faculty of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, Lima 15021, Peru
| | - Zoyla-Mirella Clavo
- Faculty of Veterinary Medicine, Universidad Nacional Mayor de San Marcos, Lima 15021, Peru
| | - Manuel Marin-Bravo
- Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima 15021, Peru
| | - Luis Inostroza-Ruiz
- Faculty of Pharmacy, Universidad Nacional Mayor de San Marcos, Lima 15021, Peru
| | | | - Miguel Quispe-Solano
- Faculty of Engineering in Food Industries, Universidad Nacional del Centro del Perú, Huancayo 12006, Peru
| | | | - Olga Palomino
- Faculty of Pharmacy, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Luis Goya
- Department of Metabolism and Nutrition, Spanish National Research Council (CSIC), Institute of Food Science, Technology and Nutrition (ICTAN), Jose Antonio Novais 10, 28040 Madrid, Spain
| |
Collapse
|
8
|
Ma MH, Li FF, Li WF, Zhao H, Jiang M, Yu YY, Dong YC, Zhang YX, Li P, Bu WJ, Sun ZJ, Dong DL. Repurposing nitazoxanide as a novel anti-atherosclerotic drug based on mitochondrial uncoupling mechanisms. Br J Pharmacol 2023; 180:62-79. [PMID: 36082580 DOI: 10.1111/bph.15949] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 08/03/2022] [Accepted: 08/28/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE The anthelmintic drug nitazoxanide has a mitochondrial uncoupling effect. Mitochondrial uncouplers have been proven to inhibit smooth muscle cell proliferation and migration, inhibit NLRP3 inflammasome activation of macrophages and improve dyslipidaemia. Therefore, we aimed to demonstrate that nitazoxanide would protect against atherosclerosis. EXPERIMENTAL APPROACH The mitochondrial oxygen consumption of cells was measured by using the high-resolution respirometry system, Oxygraph-2K. The proliferation and migration of A10 cells were measured by using Edu immunofluorescence staining, wound-induced migration and the Boyden chamber assay. Protein levels were measured by using the western blot technique. ApoE (-/-) mice were fed with a Western diet to establish an atherosclerotic model in vivo. KEY RESULTS The in vitro experiments showed that nitazoxanide and tizoxanide had a mitochondrial uncoupling effect and activated cellular AMPK. Nitazoxanide and tizoxanide inhibited serum- and PDGF-induced proliferation and migration of A10 cells. Nitazoxanide and tizoxanide inhibited NLRP3 inflammasome activation in RAW264.7 macrophages, the mechanism by which involved the AMPK/IκBα/NF-κB pathway. Nitazoxanide and tizoxanide also induced autophagy in A10 cells and RAW264.7 macrophages. The in vivo experiments demonstrated that oral administration of nitazoxanide reduced the increase in serum IL-1β and IL-6 levels and suppressed atherosclerosis in Western diet-fed ApoE (-/-) mice. CONCLUSION AND IMPLICATIONS Nitazoxanide inhibits the formation of atherosclerotic plaques in ApoE (-/-) mice fed on a Western diet. In view of nitazoxanide being an antiprotozoal drug already approved by the FDA, we propose it as a novel anti-atherosclerotic drug with clinical translational potential.
Collapse
Affiliation(s)
- Ming-Hui Ma
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Feng-Feng Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Wen-Feng Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Hui Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Man Jiang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yuan-Yuan Yu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yan-Chao Dong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Yi-Xin Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Ping Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Wen-Jie Bu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China
| | - Zhi-Jie Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| | - De-Li Dong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, People's Republic of China.,Department of Pharmacology, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
9
|
Sulodexide Increases Glutathione Synthesis and Causes Pro-Reducing Shift in Glutathione-Redox State in HUVECs Exposed to Oxygen–Glucose Deprivation: Implication for Protection of Endothelium against Ischemic Injury. Molecules 2022; 27:molecules27175465. [PMID: 36080234 PMCID: PMC9457652 DOI: 10.3390/molecules27175465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Sulodexide (SDX), a purified glycosaminoglycan mixture used to treat vascular diseases, has been reported to exert endothelial protective effects against ischemic injury. However, the mechanisms underlying these effects remain to be fully elucidated. The emerging evidence indicated that a relatively high intracellular concentration of reduced glutathione (GSH) and a maintenance of the redox environment participate in the endothelial cell survival during ischemia. Therefore, the aim of the present study was to examine the hypothesis that SDX alleviates oxygen–glucose deprivation (OGD)-induced human umbilical endothelial cells’ (HUVECs) injury, which serves as the in vitro model of ischemia, by affecting the redox state of the GSH: glutathione disulfide (GSSG) pool. The cellular GSH, GSSG and total glutathione (tGSH) concentrations were measured by colorimetric method and the redox potential (ΔEh) of the GSSG/2GSH couple was calculated, using the Nernst equation. Furthermore, the levels of the glutamate–cysteine ligase catalytic subunit (GCLc) and the glutathione synthetase (GSS) proteins, a key enzyme for de novo GSH synthesis, were determined using enzyme-linked immunoassay (ELISA). We demonstrated that the SDX treatment in OGD conditions significantly elevated the intracellular GSH, enhanced the GSH:GSSG ratio, shifting the redox potential to a more pro-reducing status. Furthermore, SDX increased the levels of both GCLc and GSS. The results show that SDX protects the human endothelial cells against ischemic stress by affecting the GSH levels and cellular redox state. These changes suggest that the reduction in the ischemia-induced vascular endothelial cell injury through repressing apoptosis and oxidative stress associated with SDX treatment may be due to an increase in GSH synthesis and modulation of the GSH redox system.
Collapse
|
10
|
Methacrylic Acid-Based Regenerative Biomaterials: Explorations into the MAAgic. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-022-00263-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
11
|
Belvedere R, Novizio N, Eletto D, Porta A, Bagnulo A, Cerciello A, Di Maio U, Petrella A. The Procoagulant Activity of Emoxilane ®: A New Appealing Therapeutic Use in Epistaxis of the Combination of Sodium Hyaluronate, Silver Salt, α-tocopherol and D-panthenol. Life (Basel) 2021; 11:life11090992. [PMID: 34575141 PMCID: PMC8472423 DOI: 10.3390/life11090992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 11/20/2022] Open
Abstract
Epistaxis is one of the most frequent hemorrhages resulting from local or systemic factors. Its management without hospitalization has prompted an interest in locally applied hemostatic agents. Generally, the therapy approaches involve sprays or creams acting as a physical barrier, even used as tampons or gauze. In this study, we have investigated the activity of Emoxilane®, a combination of sodium hyaluronate, silver salt, α-tocopherol acetate and D-panthenol, which is known to be able to separately act in a different biological manner. Our in vitro results, obtained on endothelial and nasal epithelial cells, have shown that the association of these molecules presented a notable antioxidant activity mainly due to the α-tocopherol and D-panthenol and a significant antimicrobial role thanks to the silver compound. Moreover, remarkable hemostatic activity was found by evaluating plasmin inhibition attributable to the sodium hyaluronate. Interestingly, on human plasma, we have confirmed that Emoxilane® strongly induced the increase of thrombin levels. These data suggest that the use of this association could represent an appealing pharmacological approach to actively induce hemostasis during epistaxis. Our future perspective will aim to the creation of a formulation for an easy topical application in the nose which is able to contrast the bleeding.
Collapse
Affiliation(s)
- Raffaella Belvedere
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (R.B.); (N.N.); (D.E.); (A.P.)
| | - Nunzia Novizio
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (R.B.); (N.N.); (D.E.); (A.P.)
| | - Daniela Eletto
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (R.B.); (N.N.); (D.E.); (A.P.)
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (R.B.); (N.N.); (D.E.); (A.P.)
| | - Antonino Bagnulo
- Neilos Srl, Via Bagnulo 95, 80063 Piano di Sorrento, Italy; (A.B.); (A.C.)
| | - Andrea Cerciello
- Neilos Srl, Via Bagnulo 95, 80063 Piano di Sorrento, Italy; (A.B.); (A.C.)
| | - Umberto Di Maio
- Shedir Pharma Group Spa, Via Bagnulo 95, 80063 Piano di Sorrento, Italy;
| | - Antonello Petrella
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (R.B.); (N.N.); (D.E.); (A.P.)
- Correspondence: ; Tel.: +39-089-969762; Fax: +39-089-969602
| |
Collapse
|
12
|
Zhang Y, Li C, Pei Y, Zheng L, Sun X, Zhao Z, Wang S. Trelagliptin ameliorates oxygen-glucose deprivation/reperfusion (OGD/R)-induced mitochondrial dysfunction and metabolic disturbance of endothelial cells. Hum Cell 2021; 34:1717-1726. [PMID: 34435315 DOI: 10.1007/s13577-021-00594-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/12/2021] [Indexed: 11/27/2022]
Abstract
Acute myocardial infarction (AMI) is a severe cardiovascular disease with high mortality. It is reported to be closely related to the mitochondrial dysfunction and metabolic disturbance on endothelial cells under a chronic hypoxic state. Significant declined mitochondrial respiration, ATP production, and metabolic changes are the main characteristics of endothelial injury in the disease. Trelagliptin is a DPP-4 inhibitor applied for the treatment of type II diabetes and has been recently reported to exert various pharmacological properties. In this investigation, we examined whether Trelagliptin possessed a protective effect against mitochondrial dysfunction and metabolic disturbance in human aortic valvular endothelial cells (HAVECs) under oxygen-glucose deprivation/reperfusion (OGD/R) conditions. We found that both the cytotoxicity and mitochondrial oxidative stress in HAVECs induced by OGD/R stimulation were greatly alleviated by Trelagliptin. In addition, the declined mitochondrial respiration and ATP production decreased secretion of cystathionine and creatine, and the increased production of triglyceride and adiponectin in OGD/R-challenged HAVECs was dramatically reversed by Trelagliptin, accompanied by the upregulated expression level of PGC-1α and CPT-1. Lastly, the AMPK pathway was observed to be significantly activated in OGD/R-challenged HAVECs by Trelagliptin treatment. After co-administration of the inhibitor of the AMPK pathway, the effects of Trelagliptin on mitochondrial function and metabolic alterations were significantly abolished. Taken together, our data indicate that Trelagliptin ameliorated OGD/R-induced mitochondrial disturbance and metabolic changes by activating the AMPK pathway.
Collapse
Affiliation(s)
- Yatong Zhang
- Department of pharmacy, Beijing Hospital; National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, China
| | - Chao Li
- Department of pharmacy, Beijing Hospital; National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, China
| | - Yifang Pei
- Department of pharmacy, Beijing Hospital; National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, China
| | - Li Zheng
- Department of Pharmacy, China Aerospace Science and Industry Corporation 731 Hospital, Beijing, 100074, China
| | - Xuelin Sun
- Department of pharmacy, Beijing Hospital; National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, China
| | - Zinan Zhao
- Department of pharmacy, Beijing Hospital; National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Key Laboratory of Assessment of Clinical Drugs Risk and Individual Application (Beijing Hospital), Beijing, 100730, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, #15 Dazhong Street, Liandu District, Lishui City, 323000, Zhejiang, China.
| |
Collapse
|
13
|
Panday S, Kar S, Kavdia M. How does ascorbate improve endothelial dysfunction? - A computational analysis. Free Radic Biol Med 2021; 165:111-126. [PMID: 33497797 DOI: 10.1016/j.freeradbiomed.2021.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/23/2020] [Accepted: 01/14/2021] [Indexed: 01/02/2023]
Abstract
Low levels of ascorbate (Asc) are observed in cardiovascular and neurovascular diseases. Asc has therapeutic potential for the treatment of endothelial dysfunction, which is characterized by a reduction in nitric oxide (NO) bioavailability and increased oxidative stress in the vasculature. However, the potential mechanisms remain poorly understood for the Asc mitigation of endothelial dysfunction. In this study, we developed an endothelial cell based computational model integrating endothelial cell nitric oxide synthase (eNOS) biochemical pathway with downstream reactions and interactions of oxidative stress, tetrahydrobiopterin (BH4) synthesis and biopterin ratio ([BH4]/[TBP]), Asc and glutathione (GSH). We quantitatively analyzed three Asc mediated mechanisms that are reported to improve/maintain endothelial cell function. The mechanisms include the reduction of •BH3 to BH4, direct scavenging of superoxide (O2•-) and peroxynitrite (ONOO-) and increasing eNOS activity. The model predicted that Asc at 0.1-100 μM concentrations improved endothelial cell NO production, total biopterin and biopterin ratio in a dose dependent manner and the extent of cellular oxidative stress. Asc increased BH4 availability and restored eNOS coupling under oxidative stress conditions. Asc at concentrations of 1-10 mM reduced O2•- and ONOO- levels and could act as an antioxidant. We predicted that glutathione peroxidase and peroxiredoxin in combination with GSH and Asc can restore eNOS coupling and NO production under oxidative stress conditions. Asc supplementation may be used as an effective therapeutic strategy when BH4 levels are depleted. This study provides detailed understanding of the mechanism responsible and the optimal cellular Asc levels for improvement in endothelial dysfunction.
Collapse
Affiliation(s)
- Sheetal Panday
- Department of Biomedical Engineering, Wayne State University, Detroit, 48202, MI, USA
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Kuwait
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, 48202, MI, USA.
| |
Collapse
|
14
|
Sutter CH, Rainwater HM, Sutter TR. Contributions of Nitric Oxide to AHR-Ligand-Mediated Keratinocyte Differentiation. Int J Mol Sci 2020; 21:ijms21165680. [PMID: 32784365 PMCID: PMC7460822 DOI: 10.3390/ijms21165680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022] Open
Abstract
Activation of the aryl hydrocarbon receptor (AHR) in normal human epidermal keratinocytes (NHEKs) accelerates keratinocyte terminal differentiation through metabolic reprogramming and reactive oxygen species (ROS) production. Of the three NOS isoforms, NOS3 is significantly increased at both the RNA and protein levels by exposure to the very potent and selective ligand of the AHR, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Inhibition of NOS with the chemical N-nitro-l-arginine methyl ester (l-NAME) reversed TCDD-induced cornified envelope formation, an endpoint of terminal differentiation, as well as the expression of filaggrin (FLG), a marker of differentiation. Conversely, exposure to the NO-donor, S-nitroso-N-acetyl-DL-penicillamine (SNAP), increased the number of cornified envelopes above control levels and augmented the levels of cornified envelopes formed in response to TCDD treatment and increased the expression of FLG. This indicates that nitric oxide signaling can increase keratinocyte differentiation and that it is involved in the AHR-mediated acceleration of differentiation. As the nitrosylation of cysteines is a mechanism by which NO affects the structure and functions of proteins, the S-nitrosylation biotin switch technique was used to measure protein S-nitrosylation. Activation of the AHR increased the S-nitrosylation of two detected proteins of about 72 and 20 kD in size. These results provide new insights into the role of NO and protein nitrosylation in the process of epithelial cell differentiation, suggesting a role of NOS in metabolic reprogramming and the regulation of epithelial cell fate.
Collapse
Affiliation(s)
- Carrie Hayes Sutter
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (H.M.R.); (T.R.S.)
- W. Harry Feinstone Center for Genomic Research, University of Memphis, Memphis, TN 38152, USA
- Correspondence:
| | - Haley M. Rainwater
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (H.M.R.); (T.R.S.)
| | - Thomas R. Sutter
- Department of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (H.M.R.); (T.R.S.)
- W. Harry Feinstone Center for Genomic Research, University of Memphis, Memphis, TN 38152, USA
- Department of Chemistry, University of Memphis, Memphis, TN 38152, USA
| |
Collapse
|
15
|
Serrano JJ, Delgado B, Medina MÁ. Control of tumor angiogenesis and metastasis through modulation of cell redox state. Biochim Biophys Acta Rev Cancer 2020; 1873:188352. [PMID: 32035101 DOI: 10.1016/j.bbcan.2020.188352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022]
Abstract
Redox reactions pervade all biology. The control of cellular redox state is essential for bioenergetics and for the proper functioning of many biological functions. This review traces a timeline of findings regarding the connections between redox and cancer. There is ample evidence of the involvement of cellular redox state on the different hallmarks of cancer. Evidence of the control of tumor angiogenesis and metastasis through modulation of cell redox state is reviewed and highlighted.
Collapse
Affiliation(s)
- José J Serrano
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain
| | - Belén Delgado
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain
| | - Miguel Ángel Medina
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, E-29071 Málaga, Spain; IBIMA (Biomedical Research Institute of Málaga), E-29071 Málaga, Spain; CIBER de Enfermedades Raras (CIBERER), E-29071 Málaga, Spain.
| |
Collapse
|
16
|
Xiao M, Lu D, Tian J, Yu Y, Zhang Q, Zhang L, Chang D. The protective effects of GLP-1 receptor agonist lixisenatide on oxygen-glucose deprivation/reperfusion (OGD/R)-induced deregulation of endothelial tube formation. RSC Adv 2020; 10:10245-10253. [PMID: 35498599 PMCID: PMC9050383 DOI: 10.1039/c9ra09959j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/25/2020] [Indexed: 01/01/2023] Open
Abstract
Acute myocardial infarction (AMI) is a complication of atherosclerosis that takes place in coronary arteries. Cardiac endothelial cells play a significant role in the pathogenesis of AMI. Oxygen-glucose deprivation/reperfusion (OGD/R) is widely used as a model to simulate AMI in vitro. Recently, antidiabetic GLP-1 receptor agonists have been shown to exert pleiotropic effects that modulate cardiovascular complications. In this study, we investigated the vascular effect of lixisenatide. We show that pre-treatment of endothelial cells with lixisenatide protected them from OGD/R-induced cytotoxicity and improved their viability. Pre-treatment with lixisenatide ameliorated OGD/R-induced ROS accumulation and disturbed endothelial tube formation. At the molecular level, lixisenatide mitigated OGD/R-induced reduced eNOS expression and NO production but further promoted the expression of the anti-oxidant regulators Nrf2 and HO-1. Mechanistically, we confirmed that the PI3K/Akt pathway is essential for mediating the effects of lixisenatide, and blockage of PI3K/Akt using the inhibitor LY294002 abolished the ameliorative effect of lixisenatide on ROS production and impaired tube formation. These data indicate that lixisenatide possesses a beneficial effect on the vasculature in a model of ischemia-induced endothelial injury. We conclude that the GLP-1 receptor agonist lixisenatide has pleiotropic properties that can modulate vascular function independent of its anti-glycemic effect. Acute myocardial infarction (AMI) is a complication of atherosclerosis that takes place in coronary arteries.![]()
Collapse
Affiliation(s)
- Mochao Xiao
- Department of Cardiology
- Fourth Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Daifeng Lu
- Department of Orthopaedics
- Fourth Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Jiali Tian
- Department of Cardiology
- Fourth Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Yang Yu
- Department of Cardiology
- Fourth Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Qin Zhang
- Department of Cardiology
- First Affiliated Hospital of Harbin Medical University
- Harbin
- China
| | - Lili Zhang
- Department of Cardiology
- The People's Hospital of Longhua
- The Affiliated Hospital of Southern Medical University
- Shenzhen
- China
| | - Dong Chang
- Department of Cardiology
- Xiamen Cardiovascular Hospital
- Xiamen University
- Xiamen
- China
| |
Collapse
|
17
|
Antioxidant Defence Systems and Oxidative Stress in Poultry Biology: An Update. Antioxidants (Basel) 2019; 8:antiox8070235. [PMID: 31336672 PMCID: PMC6680731 DOI: 10.3390/antiox8070235] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Poultry in commercial settings are exposed to a range of stressors. A growing body of information clearly indicates that excess ROS/RNS production and oxidative stress are major detrimental consequences of the most common commercial stressors in poultry production. During evolution, antioxidant defence systems were developed in poultry to survive in an oxygenated atmosphere. They include a complex network of internally synthesised (e.g., antioxidant enzymes, (glutathione) GSH, (coenzyme Q) CoQ) and externally supplied (vitamin E, carotenoids, etc.) antioxidants. In fact, all antioxidants in the body work cooperatively as a team to maintain optimal redox balance in the cell/body. This balance is a key element in providing the necessary conditions for cell signalling, a vital process for regulation of the expression of various genes, stress adaptation and homeostasis maintenance in the body. Since ROS/RNS are considered to be important signalling molecules, their concentration is strictly regulated by the antioxidant defence network in conjunction with various transcription factors and vitagenes. In fact, activation of vitagenes via such transcription factors as Nrf2 leads to an additional synthesis of an array of protective molecules which can deal with increased ROS/RNS production. Therefore, it is a challenging task to develop a system of optimal antioxidant supplementation to help growing/productive birds maintain effective antioxidant defences and redox balance in the body. On the one hand, antioxidants, such as vitamin E, or minerals (e.g., Se, Mn, Cu and Zn) are a compulsory part of the commercial pre-mixes for poultry, and, in most cases, are adequate to meet the physiological requirements in these elements. On the other hand, due to the aforementioned commercially relevant stressors, there is a need for additional support for the antioxidant system in poultry. This new direction in improving antioxidant defences for poultry in stress conditions is related to an opportunity to activate a range of vitagenes (via Nrf2-related mechanisms: superoxide dismutase, SOD; heme oxygenase-1, HO-1; GSH and thioredoxin, or other mechanisms: Heat shock protein (HSP)/heat shock factor (HSP), sirtuins, etc.) to maximise internal AO protection and redox balance maintenance. Therefore, the development of vitagene-regulating nutritional supplements is on the agenda of many commercial companies worldwide.
Collapse
|
18
|
Bubb KJ, Drummond GR, Figtree GA. New opportunities for targeting redox dysregulation in cardiovascular disease. Cardiovasc Res 2019; 116:532-544. [DOI: 10.1093/cvr/cvz183] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/02/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Abstract
Despite substantial promise, the use of antioxidant therapy to improve cardiovascular outcomes has been disappointing. Whilst the fundamental biology supporting their use continues to build, the challenge now is to differentially target dysregulated redox signalling domains and to identify new ways to deliver antioxidant substances. Looking further afield to other disciplines, there is an emerging ‘tool-kit’ containing sophisticated molecular and drug delivery applications. Applying these to the cardiovascular redox field could prove a successful strategy to combat the increasing disease burden. Excessive reactive oxygen species production and protein modifications in the mitochondria has been the target of successful drug development with several positive outcomes emerging in the cardiovascular space, harnessing both improved delivery mechanisms and enhanced understanding of the biological abnormalities. Using this as a blueprint, similar strategies could be applied and expanded upon in other redox-hot-spots, such as the caveolae sub-cellular region, which houses many of the key cardiovascular redox proteins such as NADPH oxidase, endothelial nitric oxide synthase, angiotensin II receptors, and beta adrenoceptors. The expanded tool kit of drug development, including gene and miRNA therapies, nanoparticle technology and micropeptide targeting, can be applied to target dysregulated redox signalling in subcellular compartments of cardiovascular cells. In this review, we consider the opportunities for improving cardiovascular outcomes by utilizing new technology platforms to target subcellular ‘bonfires’ generated by dysregulated redox pathways, to improve clinical outcomes.
Collapse
Affiliation(s)
- Kristen J Bubb
- Cardiothoracic and Vascular Health, Kolling Institute and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology and Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Australia
| | - Gemma A Figtree
- Cardiothoracic and Vascular Health, Kolling Institute and Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
19
|
Chen P, Liu H, Xiang H, Zhou J, Zeng Z, Chen R, Zhao S, Xiao J, Shu Z, Chen S, Lu H. Palmitic acid-induced autophagy increases reactive oxygen species via the Ca 2+/PKCα/NOX4 pathway and impairs endothelial function in human umbilical vein endothelial cells. Exp Ther Med 2019; 17:2425-2432. [PMID: 30906429 PMCID: PMC6425131 DOI: 10.3892/etm.2019.7269] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022] Open
Abstract
It is well known that the lipotoxic mechanism of palmitic acid (PA), a main constituent of triglyceride, is dependent on reactive oxygen species (ROS). Recently, it has also been reported that PA is an autophagy inducer. However, the causal association and underlying mechanism of induced autophagy and ROS in PA toxicity remain unclear. The present study demonstrates for the first time that PA-induced autophagy enhances ROS generation via activating the calcium ion/protein kinase Cα/nicotinamide adenine dinucleotide phosphate oxidase 4 (Ca2+/PKCα/NOX4) pathway in human umbilical vein endothelial cells (HUVECs). It was revealed that PA treatment resulted in a significant increase in ROS generation and autophagic activity, leading to endothelial dysfunction as indicated by downregulated nitric oxide synthesis, decreased capillary-like structure formation and damaged cell repair capability. Furthermore, PA effectively activated the Ca2+/PKCα/NOX4 pathway, which is indicative of upregulated cytosolic Ca2+ levels, activated PKCα and increased NOX4 protein expression. 3-Methyladenine was then used to inhibit autophagy, which significantly reduced PA-induced ROS generation and blocked the Ca2+/PKCα/NOX4 pathway. The endothelial dysfunction caused by PA was ameliorated by downregulating ROS generation using a NOX4 inhibitor. In conclusion, PA-induced autophagy contributes to endothelial dysfunction by increasing oxidative stress via the Ca2+/PKCα/NOX4 pathway in HUVECs.
Collapse
Affiliation(s)
- Pan Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hengdao Liu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jianda Zhou
- Department of Burns, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhengpeng Zeng
- Respiratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Ruifang Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shaoli Zhao
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Xiao
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhihao Shu
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shuhua Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Biochemistry, School of Life Sciences of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
20
|
Measurement of Reactive Oxygen Species (ROS) and Mitochondrial ROS in AMPK Knockout Mice Blood Vessels. Methods Mol Biol 2018; 1732:507-517. [PMID: 29480496 DOI: 10.1007/978-1-4939-7598-3_32] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Reactive oxygen species (ROS) are a group of unstable and highly reactive molecules or free radicals typically generated as by-products of cellular processes involving molecular oxygen. In vascular cells, the excessive ROS generation results in the initiation and progression of cardiovascular diseases (CVD). Therefore, a dynamic, robust, and accurate ROS detection method in the blood vessels is essential for pathophysiological research studies of the cardiovascular system.In this chapter, we describe a fluorescence dye-based detection method for assaying superoxide and mitochondrial superoxide in mouse aorta using dihydroethidium (DHE) and MitoSOX. The protocol includes preparation of frozen aortic tissue sections, monitoring DHE oxidation-derived fluorescence by fluorescence microscopy, and high-performance liquid chromatograph-based analysis of MitoSOX and its oxidation products. For studying the role of AMP-activated protein kinase (AMPK) in the redox regulation, we employed AMPKα2 knockout mice and observed increased superoxide and mitochondrial superoxide levels in the aorta of AMPK knockout mice relative to the wild-type group. This novel ROS detection method will be valuable for investigating the roles of cellular and/or mitochondrial ROS in the pathogenesis of CVDs.
Collapse
|
21
|
Panieri E, Millia C, Santoro MM. Real-time quantification of subcellular H 2O 2 and glutathione redox potential in living cardiovascular tissues. Free Radic Biol Med 2017; 109:189-200. [PMID: 28192232 DOI: 10.1016/j.freeradbiomed.2017.02.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/31/2017] [Accepted: 02/08/2017] [Indexed: 12/17/2022]
Abstract
Detecting and measuring the dynamic redox events that occur in vivo is a prerequisite for understanding the impact of oxidants and redox events in normal and pathological conditions. These aspects are particularly relevant in cardiovascular tissues wherein alterations of the redox balance are associated with stroke, aging, and pharmacological intervention. An ambiguous aspect of redox biology is how redox events occur in subcellular organelles including mitochondria, and nuclei. Genetically-encoded Rogfp2 fluorescent probes have become powerful tools for real-time detection of redox events. These probes detect hydrogen peroxide (H2O2) levels and glutathione redox potential (EGSH), both with high spatiotemporal resolution. By generating novel transgenic (Tg) zebrafish lines that express compartment-specific Rogfp2-Orp1 and Grx1-Rogfp2 sensors we analyzed cytosolic, mitochondrial, and the nuclear redox state of endothelial cells and cardiomyocytes of living zebrafish embryos. We provide evidence for the usefulness of these Tg lines for pharmacological compounds screening by addressing the blocking of pentose phosphate pathways (PPP) and glutathione synthesis, thus altering subcellular redox state in vivo. Rogfp2-based transgenic zebrafish lines represent valuable tools to characterize the impact of redox changes in living tissues and offer new opportunities for studying metabolic driven antioxidant response in biomedical research.
Collapse
Affiliation(s)
- Emiliano Panieri
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | - Carlo Millia
- Laboratory of Endothelial Molecular Biology, Vesalius Research Center, Department of Oncology, VIB-KUL, Leuven, Belgium
| | - Massimo M Santoro
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Laboratory of Endothelial Molecular Biology, Vesalius Research Center, Department of Oncology, VIB-KUL, Leuven, Belgium.
| |
Collapse
|
22
|
Cesselli D, Aleksova A, Sponga S, Cervellin C, Di Loreto C, Tell G, Beltrami AP. Cardiac Cell Senescence and Redox Signaling. Front Cardiovasc Med 2017; 4:38. [PMID: 28612009 PMCID: PMC5447053 DOI: 10.3389/fcvm.2017.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022] Open
Abstract
Aging is characterized by a progressive loss of the ability of the organism to cope with stressors and to repair tissue damage. As a result, chronic diseases, including cardiovascular disease, increase their prevalence with aging, underlining the existence of common mechanisms that lead to frailty and age-related diseases. In this frame, the progressive decline of the homeostatic and reparative function of primitive cells has been hypothesized to play a major role in the evolution of cardiac pathology to heart failure. Although initially it was believed that reactive oxygen species (ROS) were produced in an unregulated manner as a byproduct of cellular metabolism, causing macromolecular damage and aging, accumulating evidence indicate the major role played by redox signaling in physiology. Aim of this review is to critically revise evidence linking ROS to cell senescence and aging and to provide evidence of the primary role played by redox signaling, with a particular emphasis on the multifunctional protein APE1/Ref in stem cell biology. Finally, we will discuss evidence supporting the role of redox signaling in cardiovascular cells.
Collapse
Affiliation(s)
| | - Aneta Aleksova
- Cardiovascular Department, Azienda Sanitaria Universitaria Integrata di Trieste, University of Trieste, Trieste, Italy
| | - Sandro Sponga
- Cardiothoracic Surgery, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | | | - Gianluca Tell
- Department of Medicine, University of Udine, Udine, Italy
| | | |
Collapse
|
23
|
Abnormal kynurenine pathway of tryptophan catabolism in cardiovascular diseases. Cell Mol Life Sci 2017; 74:2899-2916. [PMID: 28314892 DOI: 10.1007/s00018-017-2504-2] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/26/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Kynurenine pathway (KP) is the primary path of tryptophan (Trp) catabolism in most mammalian cells. The KP generates several bioactive catabolites, such as kynurenine (Kyn), kynurenic acid (KA), 3-hydroxykynurenine (3-HK), xanthurenic acid (XA), and 3-hydroxyanthranilic acid (3-HAA). Increased catabolite concentrations in serum are associated with several cardiovascular diseases (CVD), including heart disease, atherosclerosis, and endothelial dysfunction, as well as their risk factors, including hypertension, diabetes, obesity, and aging. The first catabolic step in KP is primarily controlled by indoleamine 2,3-dioxygenase (IDO) and tryptophan 2,3-dioxygenase (TDO). Following this first step, the KP has two major branches, one branch is mediated by kynurenine 3-monooxygenase (KMO) and kynureninase (KYNU) and is responsible for the formation of 3-HK, 3-HAA, and quinolinic acid (QA); and another branch is controlled by kynurenine amino-transferase (KAT), which generates KA. Uncontrolled Trp catabolism has been demonstrated in distinct CVD, thus, understanding the underlying mechanisms by which regulates KP enzyme expression and activity is paramount. This review highlights the recent advances on the effect of KP enzyme expression and activity in different tissues on the pathological mechanisms of specific CVD, KP is an inflammatory sensor and modulator in the cardiovascular system, and KP catabolites act as the potential biomarkers for CVD initiation and progression. Moreover, the biochemical features of critical KP enzymes and principles of enzyme inhibitor development are briefly summarized, as well as the therapeutic potential of KP enzyme inhibitors against CVD is briefly discussed.
Collapse
|
24
|
Vochysia rufa Stem Bark Extract Protects Endothelial Cells against High Glucose Damage. MEDICINES 2017; 4:medicines4010009. [PMID: 28930225 PMCID: PMC5597076 DOI: 10.3390/medicines4010009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/27/2017] [Accepted: 02/15/2017] [Indexed: 01/26/2023]
Abstract
Background: Increased oxidative stress by persistent hyperglycemia is a widely accepted factor in vascular damage responsible for type 2 diabetes complications. The plant Vochysia rufa (Vr) has been used in folk medicine in Brazil for the treatment of diabetes. Thus; the protective effect of a Vr stem bark extract against a challenge by a high glucose concentration on EA.hy926 (EA) endothelial cells is evaluated. Methods: Vegetal material is extracted with distilled water by maceration and evaporated until dryness under vacuum. Then; it is isolated by capillary electrophoresis-tandem mass spectrometry. Cell viability is evaluated on EA cells treated with 0.5-100 µg/mL of the Vr extract for 24 h. The extract is diluted at concentrations of 5, 10 and 25 µg/mL and maintained for 24 h along with 30 mM of glucose to evaluate its protective effect on reduced glutathione (GSH); glutathione peroxidase (GPx) and reductase (GR) and protein carbonyl groups. Results:V. rufa stem bark is composed mainly of sugars; such as inositol; galactose; glucose; mannose; sacarose; arabinose and ribose. Treatment with Vr up to 100 µg/mL for 24 h did not affect cell viability. Treatment of EA cells with 30 mM of glucose for 24 h significantly increased the cell damage. EA cells treated with 30 mM of glucose showed a decrease of GSH concentration and increased Radical Oxygen Species (ROS) and activity of antioxidant enzymes and protein carbonyl levels; compared to control. Co-treatment of EA with 30 mM glucose plus 1-10 μg/mL Vr significantly reduced cell damage while 5-25 μg/mL Vr evoked a significant protection against the glucose insult; recovering ROS; GSH; antioxidant enzymes and carbonyls to baseline levels. Conclusion:V. rufa extract protects endothelial cells against oxidative damage by modulating ROS; GSH concentration; antioxidant enzyme activity and protein carbonyl levels.
Collapse
|
25
|
Förstermann U, Xia N, Li H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ Res 2017; 120:713-735. [DOI: 10.1161/circresaha.116.309326] [Citation(s) in RCA: 692] [Impact Index Per Article: 98.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/19/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
Major reactive oxygen species (ROS)–producing systems in vascular wall include NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, the mitochondrial electron transport chain, and uncoupled endothelial nitric oxide (NO) synthase. ROS at moderate concentrations have important signaling roles under physiological conditions. Excessive or sustained ROS production, however, when exceeding the available antioxidant defense systems, leads to oxidative stress. Animal studies have provided compelling evidence demonstrating the roles of vascular oxidative stress and NO in atherosclerosis. All established cardiovascular risk factors such as hypercholesterolemia, hypertension, diabetes mellitus, and smoking enhance ROS generation and decrease endothelial NO production. Key molecular events in atherogenesis such as oxidative modification of lipoproteins and phospholipids, endothelial cell activation, and macrophage infiltration/activation are facilitated by vascular oxidative stress and inhibited by endothelial NO. Atherosclerosis develops preferentially in vascular regions with disturbed blood flow (arches, branches, and bifurcations). The fact that these sites are associated with enhanced oxidative stress and reduced endothelial NO production is a further indication for the roles of ROS and NO in atherosclerosis. Therefore, prevention of vascular oxidative stress and improvement of endothelial NO production represent reasonable therapeutic strategies in addition to the treatment of established risk factors (hypercholesterolemia, hypertension, and diabetes mellitus).
Collapse
Affiliation(s)
- Ulrich Förstermann
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Ning Xia
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Huige Li
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| |
Collapse
|
26
|
Wang Q, Zhang M, Torres G, Wu S, Ouyang C, Xie Z, Zou MH. Metformin Suppresses Diabetes-Accelerated Atherosclerosis via the Inhibition of Drp1-Mediated Mitochondrial Fission. Diabetes 2017; 66:193-205. [PMID: 27737949 PMCID: PMC5204316 DOI: 10.2337/db16-0915] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/06/2016] [Indexed: 12/21/2022]
Abstract
Metformin is a widely used antidiabetic drug that exerts cardiovascular protective effects in patients with diabetes. How metformin protects against diabetes-related cardiovascular diseases remains poorly understood. Here, we show that metformin abated the progression of diabetes-accelerated atherosclerosis by inhibiting mitochondrial fission in endothelial cells. Metformin treatments markedly reduced mitochondrial fragmentation, mitigated mitochondrial-derived superoxide release, improved endothelial-dependent vasodilation, inhibited vascular inflammation, and suppressed atherosclerotic lesions in streptozotocin (STZ)-induced diabetic ApoE-/- mice. In high glucose-exposed endothelial cells, metformin treatment and adenoviral overexpression of constitutively active AMPK downregulated mitochondrial superoxide, lowered levels of dynamin-related protein (Drp1) and its translocation into mitochondria, and prevented mitochondrial fragmentation. In contrast, AMPK-α2 deficiency abolished the effects of metformin on Drp1 expression, oxidative stress, and atherosclerosis in diabetic ApoE-/-/AMPK-α2-/- mice, indicating that metformin exerts an antiatherosclerotic action in vivo via the AMPK-mediated blockage of Drp1-mediated mitochondrial fission. Consistently, mitochondrial division inhibitor 1, a potent and selective Drp1 inhibitor, reduced mitochondrial fragmentation, attenuated oxidative stress, ameliorated endothelial dysfunction, inhibited inflammation, and suppressed atherosclerosis in diabetic mice. These findings show that metformin attenuated the development of atherosclerosis by reducing Drp1-mediated mitochondrial fission in an AMPK-dependent manner. Suppression of mitochondrial fission may be a therapeutic approach for treating macrovascular complications in patients with diabetes.
Collapse
Affiliation(s)
- Qilong Wang
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Miao Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Gloria Torres
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Shengnan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Changhan Ouyang
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Zhonglin Xie
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
- Corresponding author: Ming-Hui Zou, , or Zhonglin Xie,
| |
Collapse
|
27
|
Karimi Galougahi K, Ashley EA, Ali ZA. Redox regulation of vascular remodeling. Cell Mol Life Sci 2016; 73:349-63. [PMID: 26483132 PMCID: PMC11108558 DOI: 10.1007/s00018-015-2068-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 01/09/2023]
Abstract
Vascular remodeling is a dynamic process of structural and functional changes in response to biochemical and biomechanical signals in a complex in vivo milieu. While inherently adaptive, dysregulation leads to maladaptive remodeling. Reactive oxygen species participate in homeostatic cell signaling in tightly regulated- and compartmentalized cellular circuits. It is well established that perturbations in oxidation-reduction (redox) homeostasis can lead to a state of oxidative-, and more recently, reductive stress. We provide an overview of the redox signaling in the vasculature and review the role of oxidative- and reductive stress in maladaptive vascular remodeling. Particular emphasis has been placed on essential processes that determine phenotype modulation, migration and fate of the main cell types in the vessel wall. Recent advances in systems biology and the translational opportunities they may provide to specifically target the redox pathways driving pathological vascular remodeling are discussed.
Collapse
Affiliation(s)
- Keyvan Karimi Galougahi
- Division of Cardiology, Center for Interventional Vascular Therapy, New York Presbyterian Hospital and Columbia University, New York, NY, USA.
- Sydney Medical School Foundation, University of Sydney, Sydney, Australia.
| | - Euan A Ashley
- Division of Cardiology, Stanford University, Stanford, CA, USA
| | - Ziad A Ali
- Division of Cardiology, Center for Interventional Vascular Therapy, New York Presbyterian Hospital and Columbia University, New York, NY, USA
- Cardiovascular Research Foundation, New York, NY, USA
| |
Collapse
|
28
|
Pulmonary Protection Strategies in Cardiac Surgery: Are We Making Any Progress? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:416235. [PMID: 26576223 PMCID: PMC4630421 DOI: 10.1155/2015/416235] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/12/2015] [Indexed: 01/19/2023]
Abstract
Pulmonary dysfunction is a common complication of cardiac surgery. The mechanisms involved in the development of pulmonary dysfunction are multifactorial and can be related to the activation of inflammatory and oxidative stress pathways. Clinical manifestation varies from mild atelectasis to severe respiratory failure. Managing pulmonary dysfunction postcardiac surgery is a multistep process that starts before surgery and continues during both the operative and postoperative phases. Different pulmonary protection strategies have evolved over the years; however, the wide acceptance and clinical application of such techniques remain hindered by the poor level of evidence or the sample size of the studies. A better understanding of available modalities and/or combinations can result in the development of customised strategies for the different cohorts of patients with the potential to hence maximise patients and institutes benefits.
Collapse
|
29
|
Wang B, Zhang Q, Yao R, Liu X, Qu Z. 7, 8-Dihydroxyflavone Protects an Endothelial Cell Line from H2O2 Damage. PLoS One 2015; 10:e0135345. [PMID: 26266800 PMCID: PMC4534426 DOI: 10.1371/journal.pone.0135345] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/22/2015] [Indexed: 12/31/2022] Open
Abstract
7, 8-dihydroxyflavone (7, 8-DHF), a selective agonist for TrkB receptors, has been well studied for its neurotrophic functions. However, its roles outside the neural tissues have scarcely been studied as yet. In this study, we investigated the protecting roles of 7, 8-DHF in EA.hy926 cells, a human umbilic vein endothelial cell line which was exposed to hydrogen peroxide (H2O2). We found that 7, 8-DHF significantly protected the cells from being damaged by H2O2 through suppression of apoptosis, attenuation of inflammatory factor releasing and inhibition of reactive oxygen species generation. The potent biological effects of 7, 8-DHF were probably executed via its binding to TrkB receptors because the receptor specific antagonist ANA-12 significantly blocked its protecting effects. The protecting roles of 7, 8-DHF in EA.hy926 cells suggest that it will be a promising compound to be developed into a health product that definitely benefits endothelial functions and prevents cardiovascular diseases.
Collapse
Affiliation(s)
- Bingxiang Wang
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China; Department of Physiology, Taishan Medical College, Taian, 271000, China
| | - Qian Zhang
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| | - Ruyong Yao
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| | - Xiangping Liu
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| | - Zhiqiang Qu
- Center for Medical Research, the First Affiliated Hospital, Qingdao University, Qingdao, 266555, China
| |
Collapse
|
30
|
Ampem PT, Smedlund K, Vazquez G. Pharmacological evidence for a role of the transient receptor potential canonical 3 (TRPC3) channel in endoplasmic reticulum stress-induced apoptosis of human coronary artery endothelial cells. Vascul Pharmacol 2015. [PMID: 26215710 DOI: 10.1016/j.vph.2015.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Unresolved endoplasmic reticulum (ER) stress, with the subsequent persistent activation of the unfolded protein response (UPR) is a well-recognized mechanism of endothelial cell apoptosis with a major impact on the integrity of the endothelium during the course of cardiovascular diseases. As in other cell types, Ca(2+) influx into endothelial cells can promote ER stress and/or contribute to mechanisms associated with it. In previous work we showed that in human coronary artery endothelial cells (HCAECs) the Ca(2+)-permeable non-selective cation channel Transient Receptor Potential Canonical 3 (TRPC3) mediates constitutive Ca(2+) influx which is critical for operation of inflammatory signaling in these cells, through a mechanism that entails coupling of TRPC3 constitutive function to activation of Ca(2+)/calmodulin-dependent protein kinase II (CAMKII). TRPC3 has been linked to UPR signaling and apoptosis in cells other than endothelial, and CAMKII is a mediator of ER stress-induced apoptosis in various cell types, including endothelial cells. In the present work we used a pharmacological approach to examine whether in HCAECs TRPC3 and CAMKII also contribute to mechanisms of ER stress-induced apoptosis. The findings show for the first time that in HCAECs activation of the UPR and the subsequent ER stress-induced apoptosis exhibit a strong requirement for constitutive Ca(2+) influx and that TRPC3 contributes to this process. In addition, we obtained evidence indicating that, similar to its roles in non-endothelial cells, CAMKII participates in ER stress-induced apoptosis in HCAECs.
Collapse
Affiliation(s)
- Prince T Ampem
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Health Science Campus, 3000 Transverse Dr., Toledo, OH 43614, USA
| | - Kathryn Smedlund
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Health Science Campus, 3000 Transverse Dr., Toledo, OH 43614, USA
| | - Guillermo Vazquez
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Health Science Campus, 3000 Transverse Dr., Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research, Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, Health Science Campus, 3000 Transverse Dr., Toledo, OH 43614, USA.
| |
Collapse
|
31
|
Silymarin as a Natural Antioxidant: An Overview of the Current Evidence and Perspectives. Antioxidants (Basel) 2015; 4:204-47. [PMID: 26785346 PMCID: PMC4665566 DOI: 10.3390/antiox4010204] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 02/06/2015] [Accepted: 03/09/2015] [Indexed: 12/16/2022] Open
Abstract
Silymarin (SM), an extract from the Silybum marianum (milk thistle) plant containing various flavonolignans (with silybin being the major one), has received a tremendous amount of attention over the last decade as a herbal remedy for liver treatment. In many cases, the antioxidant properties of SM are considered to be responsible for its protective actions. Possible antioxidant mechanisms of SM are evaluated in this review. (1) Direct scavenging free radicals and chelating free Fe and Cu are mainly effective in the gut. (2) Preventing free radical formation by inhibiting specific ROS-producing enzymes, or improving an integrity of mitochondria in stress conditions, are of great importance. (3) Maintaining an optimal redox balance in the cell by activating a range of antioxidant enzymes and non-enzymatic antioxidants, mainly via Nrf2 activation is probably the main driving force of antioxidant (AO) action of SM. (4) Decreasing inflammatory responses by inhibiting NF-κB pathways is an emerging mechanism of SM protective effects in liver toxicity and various liver diseases. (5) Activating vitagenes, responsible for synthesis of protective molecules, including heat shock proteins (HSPs), thioredoxin and sirtuins and providing additional protection in stress conditions deserves more attention. (6) Affecting the microenvironment of the gut, including SM-bacteria interactions, awaits future investigations. (7) In animal nutrition and disease prevention strategy, SM alone, or in combination with other hepatho-active compounds (carnitine, betaine, vitamin B12, etc.), might have similar hepatoprotective effects as described in human nutrition.
Collapse
|